Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 14(11): 770, 2023 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-38007517

RESUMEN

Phenotypic and functional changes in vascular smooth muscle cells (VSMCs) contribute significantly to cardiovascular diseases (CVD) but factors driving early adverse vascular changes are poorly understood. We report on novel and important roles for the Brn-3b/POU4F2 (Brn-3b) transcription factor (TF) in controlling VSMC integrity and function. Brn-3b protein is expressed in mouse aorta with localisation to VSMCs. Male Brn-3b knock-out (KO) aortas displayed extensive remodelling with increased extracellular matrix (ECM) deposition, elastin fibre disruption and small but consistent narrowing/coarctation in the descending aortas. RNA sequencing analysis showed that these effects were linked to deregulation of genes required for calcium (Ca2+) signalling, vascular contractility, sarco-endoplasmic reticulum (S/ER) stress responses and immune function in Brn-3b KO aortas and validation studies confirmed changes in Ca2+ signalling genes linked to increased intracellular Ca2+ and S/ER Ca2+ depletion [e.g. increased, Cacna1d Ca2+ channels; ryanodine receptor 2, (RyR2) and phospholamban (PLN) but reduced ATP2a1, encoding SERCA1 pump] and chaperone proteins, Hspb1, HspA8, DnaJa1 linked to increased S/ER stress, which also contributes to contractile dysfunction. Accordingly, vascular rings from Brn-3b KO aortas displayed attenuated contractility in response to KCl or phenylephrine (PE) while Brn-3b KO-derived VSMC displayed abnormal Ca2+ signalling following ATP stimulation. This data suggests that Brn-3b target genes are necessary to maintain vascular integrity /contractile function and deregulation upon loss of Brn-3b will contribute to contractile dysfunction linked to CVD.


Asunto(s)
Enfermedades Cardiovasculares , Músculo Liso Vascular , Animales , Masculino , Ratones , Aorta/metabolismo , Calcio/metabolismo , Enfermedades Cardiovasculares/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factor de Transcripción Brn-3B/metabolismo
2.
Exp Eye Res ; 213: 108806, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34715090

RESUMEN

Glaucoma is the second leading cause of blindness worldwide. This multifactorial, neurodegenerative group of diseases is characterized by the progressive loss of retinal ganglion cells (RGCs) and their axons, leading to irreversible visual impairment and blindness. There is a huge unmet and urging need for the development of new and translatable strategies and treatment options to prevent this progressive loss of RGC. Accumulating evidence points towards a critical role of neuroinflammation, in particular microglial cells, in the pathogenesis of glaucoma. Leukotrienes are mediators of neuroinflammation and are involved in many neurodegenerative diseases. Therefore, we tested the leukotriene receptors CysLT1R/GPR17-selective antagonist Montelukast (MTK) for its efficacy to modulate the reactive state of microglia in order to ameliorate RGCs loss in experimental glaucoma. Ocular hypertension (OHT) was induced unilaterally by injection of 8 µm magnetic microbead (MB) into the anterior chamber of female Brown Norway rats. The contralateral, untreated eye served as control. Successful induction of OHT was verified by daily IOP measurement using a TonoLab rebound tonometer. Simultaneously to OHT induction, one group received daily MTK treatment and the control group vehicle solution by oral gavage. Animals were sacrificed 13-15 days after MB injection. Retina and optic nerves (ON) of OHT and contralateral eyes were analyzed by immunofluorescence with specific markers for RGCs (Brn3a), microglial cells/macrophages (Iba1 and CD68), and cysteinyl leukotriene pathway receptors (CysLT1R and GPR17). Protein labeling was documented by confocal microscopy and analyzed with ImageJ plugins. Further, mRNA expression of genes of the inflammatory and leukotriene pathway was analyzed in retinal tissue. MTK treatment resulted in a short-term IOP reduction at day 2, which dissipated by day 5 of OHT induction in MTK treated animals. Furthermore, MTK treatment resulted in a decreased activation of Iba1+ microglial cells in the retina and ON, and in a significantly increased RGC survival in OHT eyes. Within the retina, GPR17 and CysLT1R expression was demonstrated in single RCGs and in microglial cells respectively. Further, increased mRNA expression of pro-inflammatory genes was detected in OHT induced retinas. In the ON, OHT induction increased the number of GPR17+ cells, showing a trend of reduction following MTK treatment. This study shows for the first time a significantly increased RGC survival in an acute OHT model following treatment with the leukotriene receptor antagonist MTK. These results strongly suggest a neuroprotective effect of MTK and a potential new therapeutic strategy for glaucoma treatment.


Asunto(s)
Antagonistas de Leucotrieno/uso terapéutico , Microglía/metabolismo , Hipertensión Ocular/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores de Leucotrienos/metabolismo , Células Ganglionares de la Retina/fisiología , Acetatos/uso terapéutico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/metabolismo , Supervivencia Celular/fisiología , Ciclopropanos/uso terapéutico , Modelos Animales de Enfermedad , Electrorretinografía , Femenino , Regulación de la Expresión Génica/fisiología , Presión Intraocular/fisiología , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Hipertensión Ocular/fisiopatología , Quinolinas/uso terapéutico , ARN Mensajero/genética , Ratas , Ratas Endogámicas BN , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/metabolismo , Retina/fisiopatología , Sulfuros/uso terapéutico , Tonometría Ocular , Factor de Transcripción Brn-3B/metabolismo
3.
Cancer Sci ; 112(10): 4176-4186, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34327778

RESUMEN

As a POU homeodomain transcription factor, POU4F2 has been implicated in regulating tumorigenic processes in various cancers. However, the role of POU4F2 in colorectal cancer (CRC) remains unclear. Here, we revealed that POU4F2 functions as a tumor promotor in CRC. Bioinformatics analysis in specimens from CRC patients and expression analysis in CRC cell lines showed that POU4F2 was upregulated at the mRNA and protein levels in CRC. Depletion of POU4F2 suppressed the metastatic phenotypes of CRC cells, including cell migration, invasion, and the expression of epithelial-mesenchymal transition (EMT) markers. Moreover, depletion of POU4F2 decreased the number of lung metastatic nodes in nude mice. Mechanistically, POU4F2 positively regulated the Hedgehog signaling pathway, as inferred from the downregulation of the expression of sonic Hedgehog homolog, patched 1, Smoothened, and GLI family zinc finger 1 in vitro and vivo following silencing of POU4F2. Furthermore, the SMO agonist SAG reversed the effects of POU4F2 knockdown in CRC. Functionally, POU4F2 contributed to the Hedgehog signaling-regulated activation of the EMT process and promotion of CRC cell migration and invasion. Collectively, these findings elucidated the role of POU4F2 as a tumor promotor in CRC through the regulation of Hedgehog signaling-mediated EMT and suggested that POU4F2 suppression might be a promising therapeutic target in inhibiting CRC metastasis.


Asunto(s)
Movimiento Celular , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Proteínas Hedgehog/metabolismo , Invasividad Neoplásica , Factor de Transcripción Brn-3B/fisiología , Animales , Línea Celular Tumoral , Colon/metabolismo , Colon/patología , Neoplasias Colorrectales/patología , Ciclohexilaminas/farmacología , Regulación hacia Abajo , Silenciador del Gen , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Terapia Molecular Dirigida , Receptor Patched-1/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Receptor Smoothened/agonistas , Receptor Smoothened/metabolismo , Tiofenos/farmacología , Factor de Transcripción Brn-3B/antagonistas & inhibidores , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Regulación hacia Arriba , Dedos de Zinc
4.
Cell Death Dis ; 12(3): 267, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712567

RESUMEN

Metabolic and cardiovascular diseases are highly prevalent and chronic conditions that are closely linked by complex molecular and pathological changes. Such adverse effects often arise from changes in the expression of genes that control essential cellular functions, but the factors that drive such effects are not fully understood. Since tissue-specific transcription factors control the expression of multiple genes, which affect cell fate under different conditions, then identifying such regulators can provide valuable insight into the molecular basis of such diseases. This review explores emerging evidence that supports novel and important roles for the POU4F2/Brn-3b transcription factor (TF) in controlling cellular genes that regulate cardiometabolic function. Brn-3b is expressed in insulin-responsive metabolic tissues (e.g. skeletal muscle and adipose tissue) and is important for normal function because constitutive Brn-3b-knockout (KO) mice develop profound metabolic dysfunction (hyperglycaemia; insulin resistance). Brn-3b is highly expressed in the developing hearts, with lower levels in adult hearts. However, Brn-3b is re-expressed in adult cardiomyocytes following haemodynamic stress or injury and is necessary for adaptive cardiac responses, particularly in male hearts, because male Brn-3b KO mice develop adverse remodelling and reduced cardiac function. As a TF, Brn-3b regulates the expression of multiple target genes, including GLUT4, GSK3ß, sonic hedgehog (SHH), cyclin D1 and CDK4, which have known functions in controlling metabolic processes but also participate in cardiac responses to stress or injury. Therefore, loss of Brn-3b and the resultant alterations in the expression of such genes could potentially provide the link between metabolic dysfunctions with adverse cardiovascular responses, which is seen in Brn-3b KO mutants. Since the loss of Brn-3b is associated with obesity, type II diabetes (T2DM) and altered cardiac responses to stress, this regulator may provide a new and important link for understanding how pathological changes arise in such endemic diseases.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Metabolismo Energético , Síndrome Metabólico/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Animales , Factores de Riesgo Cardiometabólico , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/patología , Sistema Cardiovascular/fisiopatología , Regulación de la Expresión Génica , Humanos , Síndrome Metabólico/epidemiología , Síndrome Metabólico/genética , Síndrome Metabólico/fisiopatología , Pronóstico , Transducción de Señal , Factor de Transcripción Brn-3B/genética
5.
Nat Commun ; 12(1): 1465, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674582

RESUMEN

Atoh7 has been believed to be essential for establishing the retinal ganglion cell (RGC) lineage, and Pou4f2 and Isl1 are known to regulate RGC specification and differentiation. Here we report our further study of the roles of these transcription factors. Using bulk RNA-seq, we identify genes regulated by the three transcription factors, which expand our understanding of the scope of downstream events. Using scRNA-seq on wild-type and mutant retinal cells, we reveal a transitional cell state of retinal progenitor cells (RPCs) co-marked by Atoh7 and other genes for different lineages and shared by all early retinal lineages. We further discover the unexpected emergence of the RGC lineage in the absence of Atoh7. We conclude that competence of RPCs for different retinal fates is defined by lineage-specific genes co-expressed in the transitional state and that Atoh7 defines the RGC competence and collaborates with other factors to shepherd transitional RPCs to the RGC lineage.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Transcriptoma , Animales , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Mutación con Pérdida de Función , Ratones , ARN Citoplasmático Pequeño , Análisis de Secuencia , Células Madre , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Cell Mol Life Sci ; 78(3): 889-907, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32965515

RESUMEN

The melanopsin-expressing, intrinsically photosensitive retinal ganglion cells (ipRGCs) are a relatively recently discovered class of atypical ganglion cell photoreceptor. These ipRGCs are a morphologically and physiologically heterogeneous population that project widely throughout the brain and mediate a wide array of visual functions ranging from photoentrainment of our circadian rhythms, to driving the pupillary light reflex to improve visual function, to modulating our mood, alertness, learning, sleep/wakefulness, regulation of body temperature, and even our visual perception. The presence of melanopsin as a unique molecular signature of ipRGCs has allowed for the development of a vast array of molecular and genetic tools to study ipRGC circuits. Given the emerging complexity of this system, this review will provide an overview of the genetic tools and methods used to study ipRGCs, how these tools have been used to dissect their role in a variety of visual circuits and behaviors in mice, and identify important directions for future study.


Asunto(s)
Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/metabolismo , Animales , Animales Modificados Genéticamente/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/ultraestructura , Opsinas de Bastones/genética , Canales Catiónicos TRPC/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Fosfolipasas de Tipo C/metabolismo , Vías Visuales/fisiología
7.
Mol Vis ; 26: 691-704, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33088173

RESUMEN

Purpose: The present study aimed to determine whether the administration of Acer palmatum thumb. leaf extract (KIOM-2015E) protects against the degeneration of rat retinal ganglion cells after ischemia/reperfusion (I/R) induced by midbrain cerebral artery occlusion (MCAO). Methods: Sprague-Dawley rats were subjected to 90 min of MCAO, which produces transient ischemia in both the retina and brain due to the use of an intraluminal filament that blocks the ophthalmic and middle cerebral arteries. This was followed by reperfusion under anesthesia with isoflurane. The day after surgery, the eyes were treated three times (eye drop) or one time (oral administration) daily with KIOM-2015E for five days. Retinal histology was assessed in flat mounts and vertical sections to determine the effect of KIOM-2015E on I/R injury. Results: A significant loss of brain-specific homeobox/POU domain protein 3A (Brn3a) and neuron-specific class III beta-tubulin (Tuj-1) fluorescence and a marked increase in glial fibrillary acidic protein (GFAP) and glutamine synthetase (GS) expression were observed after five days in the PBS-treated MCAO group compared to the sham-operated control group. However, KIOM-2015E treatment reduced (1) MCAO-induced upregulation of GFAP and GS, (2) retinal ganglion cell loss, (3) nerve fiber degeneration, and (4) the number of TUNEL-positive cells. KIOM-2015E application also increased staining for parvalbumin (a marker of horizontal cell associated calcium-binding protein and amacrine cells) and recoverin (a marker of photoreceptor expression) in rats subjected to MCAO-induced retinal damage. Conclusions: Our findings indicated that KIOM-2015E treatment exerted protective effects against retinal damage following MCAO injury and that this extract may aid in the development of novel therapeutic strategies for retinal diseases, such as glaucoma and age-related macular disease.


Asunto(s)
Acer/metabolismo , Apoptosis/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Daño por Reperfusión/metabolismo , Degeneración Retiniana/prevención & control , Células Ganglionares de la Retina/efectos de los fármacos , Acer/química , Animales , Cromatografía Líquida de Alta Presión , Regulación hacia Abajo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Masculino , Fibras Nerviosas/patología , Hojas de la Planta/química , Hojas de la Planta/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/complicaciones , Daño por Reperfusión/mortalidad , Degeneración Retiniana/complicaciones , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/patología , Factor de Transcripción Brn-3B/metabolismo , Tubulina (Proteína)/metabolismo , Regulación hacia Arriba
8.
Dev Dyn ; 249(12): 1514-1528, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32741043

RESUMEN

BACKGROUND: During development, all retinal cell types arise from retinal progenitor cells (RPCs) in a step-wise fashion. Atoh7 and Pou4f2 mark, and function in, two phases of retinal ganglion cell (RGC) genesis; Atoh7 functions in a subpopulation of RPCs to render them competent for the RGC fate, whereas Pou4f2 participates in RGC fate specification and RGC differentiation. Despite extensive research on their roles, the properties of the two phases represented by these two factors have not been well studied, likely due to the retinal cellular heterogeneity. RESULTS: In this report, we describe two novel knock-in mouse alleles, Atoh7zsGreenCreERT2 and Pou4f2FlagtdTomato , which labeled retinal cells in the two phases of RGC development by fluorescent proteins. Also, the Atoh7zsGreenCreERT2 allele allowed for indirect labeling of RGCs and other cell types upon tamoxifen induction in a dose-dependent manner. Further, these alleles could be used to purify retinal cells in the different phases by fluorescence assisted cell sorting (FACS). Single cell RNA-seq analysis of purified cells from Atoh7zsGreenCreERT2 retinas further validated that this allele labeled both transitional/competent RPCs and their progenies including RGCs. CONCLUSIONS: Thus, these two alleles are very useful tools for studying the molecular and genetic mechanisms underlying RGC formation.


Asunto(s)
Alelos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/genética , Retina/embriología , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Factor de Transcripción Brn-3B/metabolismo
9.
Invest Ophthalmol Vis Sci ; 61(2): 14, 2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32049344

RESUMEN

Purpose: Experimental access to specific cell subtypes is essential for deciphering the complexity of retinal networks. Here, we characterized the selective labeling, caused by ectopic transgene expression, of two atypical retinal neurons in the ChAT-Channelrhodopsin-2 (ChR2)-EYFP mouse. Methods: Retinal sections and flat-mounts were prepared for double-staining immunohistochemistry with antibodies against EYFP and various neuronal markers. Sagittal/coronal brain slices were made to visualize EYFP signals in central nuclei. Whole-cell recordings were conducted to test the functionality of ChR2. Results: Two populations of EYFP-positive retinal cells were observed. The inner nuclear layer (INL)-located one (type I cell) distributed regularly throughout the entire retina, whereas the ganglion cell layer (GCL)-residing one (type II cell) was restricted ventrally. None of them was cholinergic, as evidenced by the complete absence of ChAT immunoreactivity. Type I cells were immunolabeled by the amacrine marker syntaxin. However, the vast majority of them were neither positive to GABA/GAD65, nor to GlyT1/glycine, suggesting that they were non-GABAergic non-glycinergic amacrine cells (nGnG ACs), which was confirmed by double-labeling with the nGnG AC marker PPP1R17. Type II cells were immunopositive to melanopsin, but not to Brn3a or Brn3b. They possessed dendrites stratifying in the outermost inner plexiform layer (IPL) and axons projecting to the suprachiasmatic nucleus (SCN) rather than the olivary pretectal nucleus (OPN), suggesting that they belonged to a Brn3b-negative subset of M1-type intrinsically photosensitive retinal ganglion cells (ipRGCs). Glutamatergic transmission-independent photocurrents were elicited in EYFP-positive cells, indicating the functional expression of ChR2. Conclusions: The ChAT-ChR2-EYFP retina exhibits ectopic, but functional, transgene expression in nGnG ACs and SCN-innervating M1 ipRGCs, thus providing an ideal tool to achieve efficient labeling and optogenetic manipulation of these cells.


Asunto(s)
Células Amacrinas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Transgenes/fisiología , Animales , Channelrhodopsins/metabolismo , Colina O-Acetiltransferasa/metabolismo , Femenino , Expresión Génica , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Transgenes/genética
10.
Nature ; 577(7790): 392-398, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31915380

RESUMEN

More than twelve morphologically and physiologically distinct subtypes of primary somatosensory neuron report salient features of our internal and external environments1-4. It is unclear how specialized gene expression programs emerge during development to endow these subtypes with their unique properties. To assess the developmental progression of transcriptional maturation of each subtype of principal somatosensory neuron, we generated a transcriptomic atlas of cells traversing the primary somatosensory neuron lineage in mice. Here we show that somatosensory neurogenesis gives rise to neurons in a transcriptionally unspecialized state, characterized by co-expression of transcription factors that become restricted to select subtypes as development proceeds. Single-cell transcriptomic analyses of sensory neurons from mutant mice lacking transcription factors suggest that these broad-to-restricted transcription factors coordinate subtype-specific gene expression programs in subtypes in which their expression is maintained. We also show that neuronal targets are involved in this process; disruption of the prototypic target-derived neurotrophic factor NGF leads to aberrant subtype-restricted patterns of transcription factor expression. Our findings support a model in which cues that emanate from intermediate and final target fields promote neuronal diversification in part by transitioning cells from a transcriptionally unspecialized state to transcriptionally distinct subtypes by modulating the selection of subtype-restricted transcription factors.


Asunto(s)
Neurogénesis , Neuronas/fisiología , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Factor de Crecimiento Nervioso/metabolismo , Neuronas/citología , ARN/análisis , ARN/genética , Análisis de la Célula Individual , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Factor de Transcripción Brn-3C/genética , Factor de Transcripción Brn-3C/metabolismo
11.
Stem Cells Dev ; 28(20): 1365-1375, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31580778

RESUMEN

Glaucoma is characterized by retinal ganglion cell (RGC) degeneration and is the second leading cause of blindness worldwide. However, current treatments such as eye drop or surgery have limitations and do not target the loss of RGC. Regenerative therapy using embryonic stem cells (ESCs) holds a promising option, but ethical concern hinders clinical applications on human subjects. In this study, we employed spermatogonial stem cells (SSCs) as an alternative source of ESCs for cell-based regenerative therapy in mouse glaucoma model. We generated functional RGCs from SSCs with a two-step protocol without applying viral transfection or chemical induction. SSCs were first dedifferentiated to embryonic stem-like cells (SSC-ESCs) that resemble ESCs in morphology, gene expression signatures, and stem cell properties. The SSC-ESCs then differentiated toward retinal lineages. We showed SSC-ESC-derived retinal cells expressed RGC-specific marker Brn3b and functioned as bona fide RGCs. To allow in vivo RGC tracing, Brn3b-EGFP reporter SSC-ESCs were generated and the derived RGCs were subsequently transplanted into the retina of glaucoma mouse models by intravitreal injection. We demonstrated that the transplanted RGCs could survive in host retina for at least 10 days after transplantation. SSC-ESC-derived RGCs can thus potentially be a novel alternative to replace the damaged RGCs in glaucomatous retina.


Asunto(s)
Células Madre Germinales Adultas/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Glaucoma/terapia , Células Ganglionares de la Retina/trasplante , Células Madre Germinales Adultas/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Expresión Génica , Genes Reporteros , Glaucoma/inducido químicamente , Glaucoma/genética , Glaucoma/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inyecciones Intravítreas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , N-Metilaspartato/administración & dosificación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Testículo/citología , Testículo/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo
12.
Development ; 146(16)2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31405994

RESUMEN

Retinal ganglion cell (RGC) degeneration is a hallmark of glaucoma, the most prevalent cause of irreversible blindness. Thus, therapeutic strategies are needed to protect and replace these projection neurons. One innovative approach is to promote de novo genesis of RGCs via manipulation of endogenous cell sources. Here, we demonstrate that the pluripotency regulator gene Krüppel-like factor 4 (Klf4) is sufficient to change the potency of lineage-restricted retinal progenitor cells to generate RGCs in vivo Transcriptome analysis disclosed that the overexpression of Klf4 induces crucial regulators of RGC competence and specification, including Atoh7 and Eya2 In contrast, loss-of-function studies in mice and zebrafish demonstrated that Klf4 is not essential for generation or differentiation of RGCs during retinogenesis. Nevertheless, induced RGCs (iRGCs) generated upon Klf4 overexpression migrate to the proper layer and project axons aligned with endogenous fascicles that reach the optic nerve head. Notably, iRGCs survive for up to 30 days after in vivo generation. We identified Klf4 as a promising candidate for reprogramming retinal cells and regenerating RGCs in the retina.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/fisiología , Neurogénesis , Células Ganglionares de la Retina/fisiología , Animales , Ciclo Celular , Femenino , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración Nerviosa , Células-Madre Neurales/fisiología , Ratas , Factor de Transcripción Brn-3A/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Pez Cebra , Proteínas de Pez Cebra/fisiología
13.
Int J Mol Sci ; 20(12)2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31197108

RESUMEN

Brn3b (Pou4f2) is a class-4 POU domain transcription factor known to play central roles in the development of different neuronal populations of the Central Nervous System, including retinal ganglion cells (RGCs), the neurons that connect the retina with the visual centers of the brain. Here, we have used CRISPR-based genetic engineering to generate a Brn3b-mCherry reporter mouse without altering the endogenous expression of Brn3b. In our mouse line, mCherry faithfully recapitulates normal Brn3b expression in the retina, the optic tracts, the midbrain tectum, and the trigeminal ganglia. The high sensitivity of mCherry also revealed novel expression of Brn3b in the neuroectodermal cells of the optic stalk during early stages of eye development. Importantly, the fluorescent intensity of Brn3b-mCherry in our reporter mice allows for noninvasive live imaging of RGCs using Scanning Laser Ophthalmoscopy (SLO), providing a novel tool for longitudinal monitoring of RGCs.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas Luminiscentes/metabolismo , Retina/metabolismo , Factor de Transcripción Brn-3B/genética , Animales , Sistemas CRISPR-Cas , Genes Reporteros , Proteínas de Homeodominio/metabolismo , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retina/diagnóstico por imagen , Factor de Transcripción Brn-3B/metabolismo , Vías Visuales/diagnóstico por imagen , Vías Visuales/metabolismo , Proteína Fluorescente Roja
14.
J Control Release ; 297: 26-38, 2019 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-30664980

RESUMEN

Glaucoma is a multifactorial neurodegenerative disorder and one of the leading causes of irreversible blindness globally and for which intraocular pressure is the only modifiable risk factor. Although neuroprotective therapies have been suggested to have therapeutic potential, drug delivery for the treatment of ocular disorders such as glaucoma remains an unmet clinical need, further complicated by poor patient compliance with topically applied treatments. In the present study we describe the development of multi-loaded PLGA-microspheres (MSs) incorporating three recognised neuroprotective agents (dexamethasone (DX), melatonin (MEL) and coenzyme Q10 (CoQ10)) in a single formulation (DMQ-MSs) to create a novel sustained-release intraocular drug delivery system (IODDS) for the treatment of glaucoma. MSs were spherical, with a mean particle size of 29.04 ±â€¯1.89 µm rendering them suitable for intravitreal injection using conventional 25G-32G needles. >62% incorporation efficiency was achieved for the three drug cargo and MSs were able to co-deliver the encapsulated active compounds in a sustained manner over 30-days with low burst release. In vitro studies showed DMQ-MSs to be neuroprotective in a glutamate-induced cytotoxicity model (IC50 10.00 ±â€¯0.94 mM versus 6.89 ±â€¯0.82 mM in absence of DMQ-MSs) in R28 cell line. In vivo efficacy studies were performed using a well-established rodent model of chronic ocular hypertension (OHT), comparing single intravitreal injections of microspheres of DMQ-MSs to their equivalent individual single-drug loaded MSs mixture (MSsmix), empty MSs, no-treatment OHT only and naïve groups. Twenty one days after OHT induction, DMQ-MSs showed a significantly neuroprotective effect on RGCs compared to OHT only controls. No such protective effect was observed in empty MSs and single-drug MSs treated groups. This work suggests that multi-loaded PLGA MSs present a novel therapeutic approach in the management of retinal neurodegeneration conditions such as glaucoma.


Asunto(s)
Portadores de Fármacos/química , Glaucoma/tratamiento farmacológico , Microesferas , Fármacos Neuroprotectores/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Animales , Dexametasona/administración & dosificación , Dexametasona/química , Modelos Animales de Enfermedad , Composición de Medicamentos , Liberación de Fármacos , Quimioterapia Combinada/métodos , Humanos , Inyecciones Intraoculares , Masculino , Melatonina/administración & dosificación , Melatonina/química , Fármacos Neuroprotectores/administración & dosificación , Ratas , Retina/efectos de los fármacos , Factor de Transcripción Brn-3B/metabolismo , Resultado del Tratamiento , Ubiquinona/administración & dosificación , Ubiquinona/análogos & derivados , Ubiquinona/química
15.
J Comp Neurol ; 526(13): 2010-2018, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29888785

RESUMEN

In addition to its well-known role in pattern vision, light influences a wide range of non-image forming, subconscious visual behaviors including circadian photoentrainment, sleep, mood, learning, and the pupillary light reflex. Each of these behaviors is thought to require input from the M1 subtype of melanopsin-expressing, intrinsically photosensitive retinal ganglion cell (ipRGC). Recent work has demonstrated that the M1 subtype of ipRGC can be further subdivided based on expression of the transcription factor Brn3b. Brn3b-positive M1 ipRGCs project to the olivary pretectal nucleus and are necessary for the pupillary light reflex, while Brn3b-negative M1 ipRGCs project to the suprachiasmatic nucleus (SCN) and are sufficient for circadian photoentrainment. However, beyond the circadian and pupil systems, little is known about the projection patterns of M1 ipRGC subtypes. Here we show that Brn3b-positive M1 ipRGCs comprise the majority of sparse M1 ipRGC inputs to the thalamus, midbrain, and hypothalamus. Our data demonstrate that very few brain targets receive convergent input from both M1 ipRGC subpopulations, suggesting that each subpopulation drives a specific subset of light-driven behaviors.


Asunto(s)
Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/biosíntesis , Animales , Ritmo Circadiano/genética , Ritmo Circadiano/fisiología , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Olivar/metabolismo , Reflejo Pupilar/fisiología , Células Ganglionares de la Retina/clasificación , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/fisiología , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo , Vías Visuales/citología
16.
Stem Cells Transl Med ; 6(11): 1972-1986, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29024560

RESUMEN

Human pluripotent stem cells have the potential to promote biological studies and accelerate drug discovery efforts by making possible direct experimentation on a variety of human cell types of interest. However, stem cell cultures are generally heterogeneous and efficient differentiation and purification protocols are often lacking. Here, we describe the generation of clustered regularly-interspaced short palindromic repeats(CRISPR)-Cas9 engineered reporter knock-in embryonic stem cell lines in which tdTomato and a unique cell-surface protein, THY1.2, are expressed under the control of the retinal ganglion cell (RGC)-enriched gene BRN3B. Using these reporter cell lines, we greatly improved adherent stem cell differentiation to the RGC lineage by optimizing a novel combination of small molecules and established an anti-THY1.2-based protocol that allows for large-scale RGC immunopurification. RNA-sequencing confirmed the similarity of the stem cell-derived RGCs to their endogenous human counterparts. Additionally, we developed an in vitro axonal injury model suitable for studying signaling pathways and mechanisms of human RGC cell death and for high-throughput screening for neuroprotective compounds. Using this system in combination with RNAi-based knockdown, we show that knockdown of dual leucine kinase (DLK) promotes survival of human RGCs, expanding to the human system prior reports that DLK inhibition is neuroprotective for murine RGCs. These improvements will facilitate the development and use of large-scale experimental paradigms that require numbers of pure RGCs that were not previously obtainable. Stem Cells Translational Medicine 2017;6:1972-1986.


Asunto(s)
Diferenciación Celular , Técnicas de Reprogramación Celular/métodos , Edición Génica/métodos , Células Madre Embrionarias Humanas/citología , Células Ganglionares de la Retina/citología , Sistemas CRISPR-Cas , Línea Celular , Células Cultivadas , Células Madre Embrionarias Humanas/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/genética , Factor de Transcripción Brn-3B/metabolismo
17.
Development ; 144(13): 2504-2516, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28676569

RESUMEN

Visual information is relayed from the eye to the brain via retinal ganglion cell (RGC) axons. Mice lacking NRP1 or NRP1-binding VEGF-A isoforms have defective RGC axon organisation alongside brain vascular defects. It is not known whether axonal defects are caused exclusively by defective VEGF-A signalling in RGCs or are exacerbated by abnormal vascular morphology. Targeted NRP1 ablation in RGCs with a Brn3bCre knock-in allele reduced axonal midline crossing at the optic chiasm and optic tract fasciculation. In contrast, Tie2-Cre-mediated endothelial NRP1 ablation induced axon exclusion zones in the optic tracts without impairing axon crossing. Similar defects were observed in Vegfa120/120 and Vegfa188/188 mice, which have vascular defects as a result of their expression of single VEGF-A isoforms. Ectopic midline vascularisation in endothelial Nrp1 and Vegfa188/188 mutants caused additional axonal exclusion zones within the chiasm. As in vitro and in vivo assays demonstrated that vessels do not repel axons, abnormally large or ectopically positioned vessels are likely to present physical obstacles to axon growth. We conclude that proper axonal wiring during brain development depends on the precise molecular control of neurovascular co-patterning.


Asunto(s)
Axones/metabolismo , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Neuropilina-1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Tipificación del Cuerpo , Diencéfalo/embriología , Diencéfalo/metabolismo , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Neovascularización Fisiológica , Quiasma Óptico/embriología , Quiasma Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Vías Visuales/metabolismo
18.
Development ; 144(9): 1698-1711, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28356311

RESUMEN

Regulated retinal ganglion cell (RGC) differentiation and axonal guidance is required for a functional visual system. Homeodomain and basic helix-loop-helix transcription factors are required for retinogenesis, as well as patterning, differentiation and maintenance of specific retinal cell types. We hypothesized that Dlx1, Dlx2 and Brn3b homeobox genes function in parallel intrinsic pathways to determine RGC fate and therefore generated Dlx1/Dlx2/Brn3b triple-knockout mice. A more severe retinal phenotype was found in the Dlx1/Dlx2/Brn3b-null retinas than was predicted by combining features of the Brn3b single- and Dlx1/Dlx2 double-knockout retinas, including near total RGC loss with a marked increase in amacrine cells in the ganglion cell layer. Furthermore, we discovered that DLX1 and DLX2 function as direct transcriptional activators of Brn3b expression. Knockdown of Dlx2 expression in primary embryonic retinal cultures and Dlx2 gain of function in utero strongly support that DLX2 is both necessary and sufficient for Brn3b expression in vivo We suggest that ATOH7 specifies RGC-committed progenitors and that Dlx1 and Dlx2 function both downstream of ATOH7 and in parallel, but cooperative, pathways that involve regulation of Brn3b expression to determine RGC fate.


Asunto(s)
Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/metabolismo , Factor de Transcripción Brn-3B/metabolismo , Factores de Transcripción/metabolismo , Vertebrados/metabolismo , Células Amacrinas/citología , Células Amacrinas/metabolismo , Animales , Apoptosis/genética , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Recuento de Células , División Celular/genética , Linaje de la Célula/genética , Proliferación Celular , Células Cultivadas , Neuronas Colinérgicas/citología , Neuronas Colinérgicas/metabolismo , Electroporación , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones Noqueados , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción Brn-3B/deficiencia , Factores de Transcripción/deficiencia
19.
J Cell Biochem ; 118(7): 1699-1711, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27935095

RESUMEN

Retinal ganglion cell layer (RGCs) is one of the important layers of retina, depleted in Glaucoma. Loss of RGC neurons is a major cellular mechanism involved in its pathogenesis resulting in severe vision loss. Stem cell therapy has emerged as a potential strategy to arrest the apoptotic loss of RGCs and also replace the degenerative cells in damaged retina. Here, we have investigated the incorporation and survival of mouse bone marrow derived Lin-ve stem cells in N-methyl-d-aspartate (NMDA)-induced mouse model of retinal degeneration. Two days after intravitreal injection of NMDA (100 mM) showed significant decrease in ganglion cell number and increase in TUNEL positive apoptotic cells in retinal layers. The injury was further characterized by immunohistochemical expression of Brn3b, GFAP, Bcl2, pCREB, CNTF, GDNF, and BDNF in retinal layers. Lin-ve cells (100,000 dose) were intravitreally transplanted after 2 days of injury and evaluated after 7, 14, and 21 days of transplantation. Transplanted cells were found to have migrated from intravitreal space and incorporated into injured retina at 7, 14, and 21 days post-transplantation. At 21 days Brn3b, CNTF, and BDNF expression was found to be upregulated whereas GDNF was downregulated when compared to respective injury time points. Molecular data showed decrease in the expression of Brn3b, BDNF, CNTF, and GDNF post transplantation when compared with injury groups. This study reveals that Lin-ve stem cells may exert neuroprotective effect in damaged retina mediated by participation of neurotrophic factors induced by stem cell transplantation at the site of injury. J. Cell. Biochem. 118: 1699-1711, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Células de la Médula Ósea/citología , N-Metilaspartato/toxicidad , Factores de Crecimiento Nervioso/metabolismo , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/metabolismo , Células Madre/citología , Animales , Células de la Médula Ósea/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Modelos Animales de Enfermedad , Femenino , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Retina/efectos de los fármacos , Retina/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología , Trasplante de Células Madre , Células Madre/fisiología , Factor de Transcripción Brn-3B/metabolismo
20.
PLoS One ; 11(6): e0157226, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27295136

RESUMEN

Pupillary light reflex (PLR) is an important clinical tool to assess the integrity of visual pathways. The available evidence suggests that melanopsin-expressing retinal ganglion cells (mRGCs) mediate PLR-driven by the classical photoreceptors (rods and cones) at low irradiances and by melanopsin activation at high irradiances. However, genetic or pharmacological elimination of melanopsin does not completely abolish PLR at high irradiances, raising the possibility that classical photoreceptors may have a role even at high irradiances. Using an inducible mouse model of photoreceptor degeneration, we asked whether classical photoreceptors are responsible for PLR at all irradiances, and found that the PLR was severely attenuated at all irradiances. Using multiple approaches, we show that the residual PLR at high irradiances in this mouse was primarily from the remnant rods and cones, with a minor contribution from melanopsin activation. In contrast, in rd1 mouse where classical photoreceptor degeneration occurs during development, the PLR was absent at low irradiances but intact at high irradiances, as reported previously. Since mRGCs receive inputs from classical photoreceptors, we also asked whether developmental loss of classical photoreceptors as in rd1 mouse leads to compensatory takeover of the high-irradiance PLR by mRGCs. Specifically, we looked at a distinct subpopulation of mRGCs that express Brn3b transcription factor, which has been shown to mediate PLR. We found that rd1 mouse had a significantly higher proportion of Brn3b-expressing M1 type of mRGCs than in the inducible model. Interestingly, inducing classical photoreceptor degeneration during development also resulted in a higher proportion of Brn3b-expressing M1 cells and partially rescued PLR at high irradiances. These results suggest that classical photoreceptors are primarily responsible for PLR at all irradiances, while melanopsin activation makes a minor contribution at very high irradiances.


Asunto(s)
Reflejo Pupilar , Células Fotorreceptoras Retinianas Conos/fisiología , Células Fotorreceptoras Retinianas Bastones/fisiología , Animales , Femenino , Proteínas de Homeodominio/análisis , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Ganglionares de la Retina/fisiología , Opsinas de Bastones/análisis , Opsinas de Bastones/metabolismo , Factor de Transcripción Brn-3B/análisis , Factor de Transcripción Brn-3B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA